1
|
Ye L, Cheng L, Deng Y, Wang S, Wu X, Ou S, Chang Q, Zhao X, Zhou W, Yu J, Wu Z. Absorption, tissue distribution, and excretion of glycycoumarin, a major bioactive coumarin from Chinese licorice ( Glycyrrhiza uralensis Fisch). Front Pharmacol 2023; 14:1216985. [PMID: 37484020 PMCID: PMC10361251 DOI: 10.3389/fphar.2023.1216985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 06/28/2023] [Indexed: 07/25/2023] Open
Abstract
Licorice (Glycyrrhiza uralensis Fisch) is a natural plant resource widely used as a food and herbal medication in China. Glycycoumarin (GCM) is a major coumarin in licorice that possesses several biological activities. However, little is known about its pharmacokinetic profile. The present study aimed to describe the oral absorption, tissue distribution, and excretion of GCM in rats. Free (parent drug) and/or total (parent drug plus the glucuronidated metabolite) GCM in biological samples was quantified before and after the hydrolysis reaction with β-glucuronidase using a reliable LC-MS/MS method. The results indicated that GCM was rapidly absorbed and transformed into its conjugated metabolites after administration. Free GCM plasma concentrations after i. v. (10 mg/kg) administration quickly decreased with an average t1/2,λz of 0.71 h, whereas the total GCM concentration reduced slowly with a t1/2, λz of 2.46 h. The area under the curve of glucuronidated metabolites was approximately four-times higher than that of free GCM. Presumably, because of hepatic and/or intestinal tract first-pass metabolism, GCM exhibited a poor bioavailability of 9.22%, as estimated from its total plasma concentration. Additionally, GCM was distributed rapidly and widely in various tissues except the brain. The liver had the highest concentration; further, GCM was promptly eliminated from test tissues after intraperitoneal (20 mg/kg) administration, but only a small amount of GCM was excreted via bile and urine. Overall, GCM is absorbed and rapidly transformed into its conjugated metabolites with low bioavailability; further, it is distributed in various tissues, except the brain. These pharmacokinetic results are helpful for better understanding the characteristics and pharmacological effects of GCM.
Collapse
Affiliation(s)
- Linhu Ye
- The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Nanhai District People’s Hospital of Foshan, Foshan, China
- Bijie City First People’s Hospital, Bijie, China
- School of Pharmacy, Zunyi Medicinal University, Zunyi, China
| | - Lei Cheng
- Bijie City First People’s Hospital, Bijie, China
| | - Yan Deng
- Bijie City First People’s Hospital, Bijie, China
| | - Sen Wang
- School of Pharmacy, Zunyi Medicinal University, Zunyi, China
| | - Xinyu Wu
- Bijie City First People’s Hospital, Bijie, China
| | - Shuiping Ou
- School of Pharmacy, Zunyi Medicinal University, Zunyi, China
| | - Qi Chang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xinqian Zhao
- Bijie City First People’s Hospital, Bijie, China
| | - Wen Zhou
- The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Nanhai District People’s Hospital of Foshan, Foshan, China
| | - Jinghua Yu
- The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Nanhai District People’s Hospital of Foshan, Foshan, China
| | - Zuqiang Wu
- The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Nanhai District People’s Hospital of Foshan, Foshan, China
| |
Collapse
|
2
|
Ye L, Fan S, Zhao P, Wu C, Liu M, Hu S, Wang P, Wang H, Bi H. Potential herb‒drug interactions between anti-COVID-19 drugs and traditional Chinese medicine. Acta Pharm Sin B 2023; 13:S2211-3835(23)00203-4. [PMID: 37360014 PMCID: PMC10239737 DOI: 10.1016/j.apsb.2023.06.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 03/21/2023] [Accepted: 04/20/2023] [Indexed: 06/28/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has spread worldwide. Effective treatments against COVID-19 remain urgently in need although vaccination significantly reduces the incidence, hospitalization, and mortality. At present, antiviral drugs including Nirmatrelvir/Ritonavir (PaxlovidTM), Remdesivir, and Molnupiravir have been authorized to treat COVID-19 and become more globally available. On the other hand, traditional Chinese medicine (TCM) has been used for the treatment of epidemic diseases for a long history. Currently, various TCM formulae against COVID-19 such as Qingfei Paidu decoction, Xuanfei Baidu granule, Huashi Baidu granule, Jinhua Qinggan granule, Lianhua Qingwen capsule, and Xuebijing injection have been widely used in clinical practice in China, which may cause potential herb-drug interactions (HDIs) in patients under treatment with antiviral drugs and affect the efficacy and safety of medicines. However, information on potential HDIs between the above anti-COVID-19 drugs and TCM formulae is lacking, and thus this work seeks to summarize and highlight potential HDIs between antiviral drugs and TCM formulae against COVID-19, and especially pharmacokinetic HDIs mediated by metabolizing enzymes and/or transporters. These well-characterized HDIs could provide useful information on clinical concomitant medicine use to maximize clinical outcomes and minimize adverse and toxic effects.
Collapse
Affiliation(s)
- Ling Ye
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Shicheng Fan
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Pengfei Zhao
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Chenghua Wu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Menghua Liu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Shuang Hu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Peng Wang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Hongyu Wang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Huichang Bi
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
3
|
Mao X, Li H, Zheng J. Effects of xenobiotics on CYP1 enzyme-mediated biotransformation and bioactivation of estradiol. Drug Metab Rev 2023; 55:1-49. [PMID: 36823774 DOI: 10.1080/03602532.2023.2177671] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
Endogenous estradiol (E2) exerts diverse physiological and pharmacological activities, commonly used for hormone replacement therapy. However, prolonged and excessive exposure to E2 potentially increases estrogenic cancer risk. Reportedly, CYP1 enzyme-mediated biotransformation of E2 is largely concerned with its balance between detoxification and carcinogenic pathways. Among the three key CYP1 enzymes (CYP1A1, CYP1A2, and CYP1B1), CYP1A1 and CYP1A2 mainly catalyze the formation of nontoxic 2-hydroxyestradiol (2-OH-E2), while CYP1B1 specifically catalyzes the formation of genotoxic 4-hydroxyestradiol (4-OH-E2). 4-OH-E2 can be further metabolized to electrophilic quinone intermediates accompanied by the generation of reactive oxygen species (ROS), triggering DNA damage. Since abnormal alterations in CYP1 activities can greatly affect the bioactivation process of E2, regulatory effects of xenobiotics on CYP1s are essential for E2-associated cancer development. To date, thousands of natural and synthetic compounds have been found to show potential inhibition and/or induction actions on the three CYP1 members. Generally, these chemicals share similar planar polycyclic skeletons, the structural motifs and substituent groups of which are important for their inhibitory/inductive efficiency and selectivity toward CYP1 enzymes. This review comprehensively summarizes these known inhibitors and/or inductors of E2-metabolizing CYP1s based on chemical categories and discusses their structure-activity relationships, which would contribute to better understanding of the correlation between xenobiotic-regulated CYP1 activities and estrogenic cancer susceptibility.
Collapse
Affiliation(s)
- Xu Mao
- Department of Pharmaceutical Analysis, College of Pharmacy, Mudanjiang Medical University, Mudanjiang, China
| | - Hui Li
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, USA
| | - Jiang Zheng
- State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang, China.,Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, China
| |
Collapse
|
4
|
Liu J, Banuvar S, Viana M, Barengolts E, Chen SN, Pauli GF, van Breemen RB. Pharmacokinetic Interactions of a Licorice Dietary Supplement with Cytochrome P450 Enzymes in Female Participants. Drug Metab Dispos 2023; 51:199-204. [PMID: 36328482 PMCID: PMC9900865 DOI: 10.1124/dmd.122.001050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 10/17/2022] [Accepted: 10/20/2022] [Indexed: 11/06/2022] Open
Abstract
Licorice, the roots and rhizomes of Glycyrrhiza glabra L., has been used as a medicinal herb, herbal adjuvant, and flavoring agent since ancient times. Recently, licorice extracts have become popular as dietary supplements used by females to alleviate menopausal symptoms. Exposure to licorice products containing high levels of glycyrrhizic acid can cause hypokalemia, but independent from this effect, preclinical data indicate that licorice can inhibit certain cytochrome P450 (P450) enzymes. To evaluate whether clinically relevant pharmacokinetic interactions of licorice with P450 enzymes exist, a phase 1 clinical investigation was carried out using a licorice extract depleted in glycyrrhizic acid (content <1%) and a cocktail containing caffeine, tolbutamide, alprazolam, and dextromethorphan, which are probe substrates for the enzymes CYP1A2, CYP2C9, CYP3A4/5, and CYP2D6, respectively. The botanically authenticated and chemically standardized extract of roots from G. glabra was consumed by 14 healthy menopausal and postmenopausal female participants twice daily for 2 weeks. The pharmacokinetics of each probe drug were evaluated immediately before and after supplementation with the licorice extract. Comparison of the average areas under the time-concentration curves (AUCs) for each probe substrate in serum showed no significant changes from licorice consumption, whereas time to reach peak concentration for caffeine and elimination half-life for tolbutamide showed small changes. According to the US Food and Drug Administration guidance, which is based on changes in the AUC of each probe substrate drug, the investigated licorice extract should not cause any clinically relevant pharmacokinetic interactions with respect to CYP3A4/5, CYP2C9, CYP2D6, or CYP1A2. SIGNIFICANCE STATEMENT: Despite generally-recognized-as-safe status, the licorice species Glycyrrhiza glabra has been associated with some toxicity. Preclinical studies suggest that G. glabra might cause pharmacokinetic drug interactions by inhibiting several cytochrome P450 enzymes. This phase 1 clinical study addressed these concerns by evaluating clinically relevant effects with respect to CYP3A4/5, CYP2C9, CYP2D6, and CYP1A2. These results showed that a standardized G. glabra extract did not cause any clinically relevant pharmacokinetic drug interactions with four major cytochrome P450 enzymes.
Collapse
Affiliation(s)
- Jialin Liu
- Linus Pauling Institute, College of Pharmacy, Oregon State University, Corvallis, Oregon (J.L., R.B.v.B.) and UIC Center for Botanical Dietary Supplements Research, Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, Illinois (S.B., M.V., E.B., S.-N.C., G.F.P., R.B.v.B.)
| | - Suzanne Banuvar
- Linus Pauling Institute, College of Pharmacy, Oregon State University, Corvallis, Oregon (J.L., R.B.v.B.) and UIC Center for Botanical Dietary Supplements Research, Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, Illinois (S.B., M.V., E.B., S.-N.C., G.F.P., R.B.v.B.)
| | - Marlos Viana
- Linus Pauling Institute, College of Pharmacy, Oregon State University, Corvallis, Oregon (J.L., R.B.v.B.) and UIC Center for Botanical Dietary Supplements Research, Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, Illinois (S.B., M.V., E.B., S.-N.C., G.F.P., R.B.v.B.)
| | - Elena Barengolts
- Linus Pauling Institute, College of Pharmacy, Oregon State University, Corvallis, Oregon (J.L., R.B.v.B.) and UIC Center for Botanical Dietary Supplements Research, Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, Illinois (S.B., M.V., E.B., S.-N.C., G.F.P., R.B.v.B.)
| | - Shao-Nong Chen
- Linus Pauling Institute, College of Pharmacy, Oregon State University, Corvallis, Oregon (J.L., R.B.v.B.) and UIC Center for Botanical Dietary Supplements Research, Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, Illinois (S.B., M.V., E.B., S.-N.C., G.F.P., R.B.v.B.)
| | - Guido F Pauli
- Linus Pauling Institute, College of Pharmacy, Oregon State University, Corvallis, Oregon (J.L., R.B.v.B.) and UIC Center for Botanical Dietary Supplements Research, Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, Illinois (S.B., M.V., E.B., S.-N.C., G.F.P., R.B.v.B.)
| | - Richard B van Breemen
- Linus Pauling Institute, College of Pharmacy, Oregon State University, Corvallis, Oregon (J.L., R.B.v.B.) and UIC Center for Botanical Dietary Supplements Research, Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, Illinois (S.B., M.V., E.B., S.-N.C., G.F.P., R.B.v.B.)
| |
Collapse
|
5
|
Cheng L, Deng Y, An L, Zhang Y, Liu H, Wu X, Wang T, Yang X, Ye L. Development and validation of an LC-MS/MS method for detection of glycyrol in rat plasma and its application to a pharmacokinetic study. Biomed Chromatogr 2023; 37:e5532. [PMID: 36269043 DOI: 10.1002/bmc.5532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 10/15/2022] [Accepted: 10/18/2022] [Indexed: 01/18/2023]
Abstract
Licorice (Glycyrrhiza uralensis) is one of the most popular edible and medicinal plants and is widely used in Asia. Glycyrol (GC) is a major coumarin present in licorice that exhibits various biological activities. We aimed to develop a highly sensitive and rapid liquid chromatography coupled with mass spectrometry method for the quantitative determination analysis of GC in rat plasma. GC showed linear calibration ranges of 1-100 and 50-2,000 ng/ml with correlation coefficients >0.99. The average extraction recovery ranged from 113.26 to 114.84%, and the relative standard deviation of internal standard normalized matrix factors ranged from 6.36 to 9.46%. The intra-day and inter-day precisions of GC were <15%, and the accuracy ranged from 95.31 to 112.72%. Pharmacokinetic studies showed that GC was distributed in the body with a volume of distribution of 9.06 L/kg, and the initial plasma concentration was 3275.11 ng/ml. The area under the plasma concentration vs. time curve was 479.25 ng h/ml. It was rapidly eliminated with a terminal elimination half-life of 1.47 h and a clearance rate of 4.24 L/h/kg. The pharmacokinetic results can help us to better understand the pharmacological effects of GC in the body.
Collapse
Affiliation(s)
- Lei Cheng
- Department of Pharmacy, the First People's Hospital of Bijie, Bijie, China
| | - Yan Deng
- Department of Pharmacy, the First People's Hospital of Bijie, Bijie, China
| | - Liqun An
- Anshun Technical College, Anshun, China
| | - Yan Zhang
- Zunyi Medicinal University, Zunyi, China
| | - Hong Liu
- Department of Pharmacy, the First People's Hospital of Bijie, Bijie, China
| | - Xinyu Wu
- Department of Pharmacy, the First People's Hospital of Bijie, Bijie, China
| | - Tingting Wang
- Department of Pharmacy, the First People's Hospital of Bijie, Bijie, China
| | - Xixiao Yang
- Department of Pharmacy, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Linhu Ye
- Department of Pharmacy, the First People's Hospital of Bijie, Bijie, China.,Zunyi Medicinal University, Zunyi, China.,Department of Pharmacy, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| |
Collapse
|
6
|
Li X, Qiu Q, Li M, Lin H, Cao S, Wang Q, Chen Z, Jiang W, Zhang W, Huang Y, Luo H, Luo L. Chemical composition and pharmacological mechanism of ephedra-glycyrrhiza drug pair against coronavirus disease 2019 (COVID-19). Aging (Albany NY) 2021; 13:4811-4830. [PMID: 33581688 PMCID: PMC7950231 DOI: 10.18632/aging.202622] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 11/30/2020] [Indexed: 12/12/2022]
Abstract
Traditional Chinese medicine (TCM) had demonstrated effectiveness in the prevention and control of COVID-19. Statistics showed that Ephedra and Glycyrrhiza were frequently used in the treatment of COVID-19. We hypothesized that the Ephedra-Glycyrrhiza drug pair is a potential choice for the treatment of COVID-19. Here, 112 active compounds were identified from Ephedra-Glycyrrhiza via network pharmacology approach. Ephedra-Glycyrrhiza pair enrichment analysis demonstrated that these compounds might participate in the cAMP, PI3K-Akt, JAK-STAT and chemokine signaling pathways, which had a high correlation with respiratory, nervous, blood circulation and digestive system-related diseases. Pathway analysis between Ephedra-Glycyrrhiza and COVID-19 showed that the key targets were TNF-α, IL2, FOS, ALB, and PTGS2. They might control PI3K-Akt signaling pathway to exert immune regulation, organ protection and antiviral effects. Molecular docking results showed that the active compounds from the Ephedra-Glycyrrhiza pair bound well to COVID-19 related targets, including the main protease (Mpro, also called 3CLpro), the spike protein (S protein), and the angiotensin-converting enzyme 2 (ACE2). The Molecular dynamics simulation was analyzed for the stability and flexibility of the complex. In conclusion, our study elucidated the potential pharmacological mechanism of Ephedra-Glycyrrhiza in the treatment of COVID-19 through multiple targets and pathways.
Collapse
Affiliation(s)
- Xiaoling Li
- Animal Experiment Center of Guangdong Medical University, Zhanjiang 524023, Guangdong, China
| | - Qin Qiu
- Graduate School of Guangdong Medical University, Zhanjiang 524023, Guangdong, China
| | - Mingyue Li
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Haowen Lin
- The First Clinical College of Guangdong Medical University, Zhanjiang 524023, Guangdong, China
| | - Shilin Cao
- Group of Sustainable Biochemical Engineering, School of Food Science and Engineering, Foshan University, Foshan 528000, Guangdong, China
- Sustainable Biochemical and Biosynthetic Engineering Center, Foshan Wu-Yuan Biotechnology Co., Ltd., Guangdong Biomedical Industrial Base, Foshan 528000, Guangdong, China
| | - Qu Wang
- The First Clinical College of Guangdong Medical University, Zhanjiang 524023, Guangdong, China
| | - Zishi Chen
- Group of Sustainable Biochemical Engineering, School of Food Science and Engineering, Foshan University, Foshan 528000, Guangdong, China
| | - Wenhao Jiang
- Group of Sustainable Biochemical Engineering, School of Food Science and Engineering, Foshan University, Foshan 528000, Guangdong, China
| | | | - Yuge Huang
- Department of Pediatrics, the Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, Guangdong, China
| | - Hui Luo
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang 524023, Guangdong, China
| | - Lianxiang Luo
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang 524023, Guangdong, China
- Marine Medical Research Institute of Zhanjiang, Zhanjiang 524023, Guangdong, China
| |
Collapse
|
7
|
Antimicrobial Effects against Oral Pathogens and Cytotoxicity of Glycyrrhiza uralensis Extract. PLANTS 2020; 9:plants9070838. [PMID: 32635264 PMCID: PMC7412329 DOI: 10.3390/plants9070838] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 06/29/2020] [Accepted: 07/01/2020] [Indexed: 11/17/2022]
Abstract
We aimed to evaluate the antimicrobial effects of Glycyrrhiza uralensis extract on Streptococcus mutans and Candida albicans and its biocompatibility for dental applications. The antimicrobial activity of the G. uralensis extracts at concentrations of 50, 100, 150, and 200 µg/mL was assessed using agar disk diffusion tests, counting the total number of colony-forming units (CFUs), spectrophotometric growth inhibitory assays, and microbial morphology observations using scanning electron microscopy (SEM; Merin, Carl Zeiss, Oberkochen, Germany). We measured the polyphenol and flavonoid contents of G. uralensis extracts using ultraviolet–visible spectrometry and the cytotoxicity of these extracts using an MTT (3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay. We identified that G. uralensis extracts had significant antimicrobial effects against S. mutans and C. albicans. The optical density of the experimental groups significantly decreased compared with that of the control group. SEM images revealed that the G. uralensis extract affected the morphology and density of S. mutans and C. albicans. The extract concentration of flavonoids, but not polyphenols, increased with increasing concentrations of the G. uralensis extract. Furthermore, cell viabilities were more than 70% for G. uralensis extracts with concentrations of 50 and 100 μg/mL. Naturally derived G. uralensis is biocompatible and exhibits an excellent antimicrobial effect against oral pathogens such as S. mutans and C. albicans. Thus, G. uralensis extracts can be used for the development of oral products that treat and prevent oral diseases.
Collapse
|
8
|
Kim Y, Shrestha R, Kim S, Kim JA, Lee J, Jeong TC, Kim JH, Lee S. In vitro characterization of glycyrol metabolites in human liver microsomes using HR-resolution MS spectrometer coupled with tandem mass spectrometry. Xenobiotica 2019; 50:380-388. [DOI: 10.1080/00498254.2019.1636418] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Younah Kim
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Riya Shrestha
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Sunjoo Kim
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, Republic of Korea
- BK21 PLUS Team for Creative Leader Program for Pharmacomics-based Future Pharmacy and Integrated Research Institute of Pharmaceutical Sciences, College of Pharmacy, The Catholic University of Korea, Bucheon, Republic of Korea
| | - Jeong Ah Kim
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Jaeick Lee
- Doping Control Center, Korea Institute of Science and Technology, Seoul, Korea
| | - Tae Cheon Jeong
- College of Pharmacy, Yeungnam University, Gyeongsan, Republic of Korea
| | - Ju-Hyun Kim
- College of Pharmacy, Yeungnam University, Gyeongsan, Republic of Korea
| | - Sangkyu Lee
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, Republic of Korea
| |
Collapse
|
9
|
Li N, Zhou T, Wu F, Wang R, Zhao Q, Zhang JQ, Yang BC, Ma BL. Pharmacokinetic mechanisms underlying the detoxification effect of Glycyrrhizae Radix et Rhizoma (Gancao): drug metabolizing enzymes, transporters, and beyond. Expert Opin Drug Metab Toxicol 2019; 15:167-177. [DOI: 10.1080/17425255.2019.1563595] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Na Li
- Department of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ting Zhou
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Fei Wu
- Engineering Research Center of Modern Preparation Technology of TCM of Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Rui Wang
- Engineering Research Center of Modern Preparation Technology of TCM of Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qing Zhao
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ji-Quan Zhang
- Engineering Research Center of Modern Preparation Technology of TCM of Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Bai-Can Yang
- Department of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Bing-Liang Ma
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
10
|
Shrestha R, Kim JH, Nam W, Lee HS, Lee JM, Lee S. Selective inhibition of CYP2C8 by fisetin and its methylated metabolite, geraldol, in human liver microsomes. Drug Metab Pharmacokinet 2018; 33:111-117. [DOI: 10.1016/j.dmpk.2017.12.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 09/20/2017] [Accepted: 12/12/2017] [Indexed: 01/09/2023]
|
11
|
Kim JH, Nam WS, Kim SJ, Kwon OK, Seung EJ, Jo JJ, Shresha R, Lee TH, Jeon TW, Ki SH, Lee HS, Lee S. Mechanism Investigation of Rifampicin-Induced Liver Injury Using Comparative Toxicoproteomics in Mice. Int J Mol Sci 2017; 18:E1417. [PMID: 28671602 PMCID: PMC5535909 DOI: 10.3390/ijms18071417] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 06/12/2017] [Accepted: 06/26/2017] [Indexed: 12/14/2022] Open
Abstract
Tuberculosis is one of the top causes of death among curable infectious diseases; it is an airborne infectious disease that killed 1.1 million people worldwide in 2010. Anti-tuberculosis drug-induced liver injury is the primary cause of drug-induced liver injury (DILI). Rifampicin is one of the most common anti-tuberculosis therapies and has well-known hepatotoxicity. To understand the mechanism of rifampicin-induced liver injury, we performed a global proteomic analysis of liver proteins by LC-MS/MS in a mouse model after the oral administration of 177 and 442.5 mg/kg rifampicin (LD10 and LD25) for 14 days. Based on the biochemical parameters in the plasma after rifampicin treatment, the hepatotoxic effect of rifampicin in the mouse liver was defined as a mixed liver injury. In the present study, we identified 1101 proteins and quantified 1038 proteins. A total of 29 and 40 proteins were up-regulated and 27 and 118 proteins were down-regulated in response to 177 and 442.5 mg/kg rifampicin, respectively. Furthermore, we performed Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses to characterize the mechanism of rifampicin-induced hepatotoxicity. In the molecular function category, glutathione transferase activity was up-regulated and proteins related to arachidonic acid metabolism were down-regulated. In the KEGG pathway enrichment-based clustering analysis, the peroxisome proliferator-activated receptor-γ (PPARγ) signaling pathway, cytochrome P450, glutathione metabolism, chemical carcinogenesis, and related proteins increased dose-dependently in rifampicin-treated livers. Taken together, this study showed in-depth molecular mechanism of rifampicin-induced liver injury by comparative toxicoproteomics approach.
Collapse
Affiliation(s)
- Ju-Hyun Kim
- BK21 PLUS Team for Creative Leader Program for Pharmacomics-Based Future Pharmacy and Integrated Research Institute of Pharmaceutical Sciences, College of Pharmacy, The Catholic University of Korea, Bucheon 14662, Korea.
| | - Woong Shik Nam
- BK21 Plus KNU Multi-Omics Based Creative Drug Research Team, College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea.
| | - Sun Joo Kim
- BK21 Plus KNU Multi-Omics Based Creative Drug Research Team, College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea.
| | - Oh Kwang Kwon
- BK21 Plus KNU Multi-Omics Based Creative Drug Research Team, College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea.
| | - Eun Ji Seung
- BK21 Plus KNU Multi-Omics Based Creative Drug Research Team, College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea.
| | - Jung Jae Jo
- BK21 Plus KNU Multi-Omics Based Creative Drug Research Team, College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea.
| | - Riya Shresha
- BK21 Plus KNU Multi-Omics Based Creative Drug Research Team, College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea.
| | - Tae Hee Lee
- Toxicological Research Center, Hoseo University, Asan 31499, Korea.
| | - Tae Won Jeon
- Toxicological Research Center, Hoseo University, Asan 31499, Korea.
| | - Sung Hwan Ki
- College of Pharmacy, Chosun University, Gwangju 61452, Korea.
| | - Hye Suk Lee
- BK21 PLUS Team for Creative Leader Program for Pharmacomics-Based Future Pharmacy and Integrated Research Institute of Pharmaceutical Sciences, College of Pharmacy, The Catholic University of Korea, Bucheon 14662, Korea.
| | - Sangkyu Lee
- BK21 Plus KNU Multi-Omics Based Creative Drug Research Team, College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea.
| |
Collapse
|